Summary

A Mouse Model of in Utero Transplantation

Published: January 27, 2011
doi:

Summary

The mouse model of in utero transplantation is a versatile tool that can be used to study the potential clinical applications of stem cell transplantation and gene therapy in the fetus. In this protocol, we present a general approach to performing this technique

Abstract

The transplantation of stem cells and viruses in utero has tremendous potential for treating congenital disorders in the human fetus. For example, in utero transplantation (IUT) of hematopoietic stem cells has been used to successfully treat patients with severe combined immunodeficiency.1,2 In several other conditions, however, IUT has been attempted without success.3 Given these mixed results, the availability of an efficient non-human model to study the biological sequelae of stem cell transplantation and gene therapy is critical to advance this field. We and others have used the mouse model of IUT to study factors affecting successful engraftment of in utero transplanted hematopoietic stem cells in both wild-type mice4-7 and those with genetic diseases.8,9 The fetal environment also offers considerable advantages for the success of in utero gene therapy. For example, the delivery of adenoviral10, adeno-associated viral10, retroviral11, and lentiviral vectors12,13 into the fetus has resulted in the transduction of multiple organs distant from the site of injection with long-term gene expression. in utero gene therapy may therefore be considered as a possible treatment strategy for single gene disorders such as muscular dystrophy or cystic fibrosis. Another potential advantage of IUT is the ability to induce immune tolerance to a specific antigen. As seen in mice with hemophilia, the introduction of Factor IX early in development results in tolerance to this protein.14

In addition to its use in investigating potential human therapies, the mouse model of IUT can be a powerful tool to study basic questions in developmental and stem cell biology. For example, one can deliver various small molecules to induce or inhibit specific gene expression at defined gestational stages and manipulate developmental pathways. The impact of these alterations can be assessed at various timepoints after the initial transplantation. Furthermore, one can transplant pluripotent or lineage specific progenitor cells into the fetal environment to study stem cell differentiation in a non-irradiated and unperturbed host environment.

The mouse model of IUT has already provided numerous insights within the fields of immunology, and developmental and stem cell biology. In this video-based protocol, we describe a step-by-step approach to performing IUT in mouse fetuses and outline the critical steps and potential pitfalls of this technique.

Protocol

1. Preparation of Injection Pipettes Calibrate the pipette puller such that separation of the glass pipette occurs within 15 seconds (see manufacturer’s instructions regarding calibration). The pipette will have a taper where it separates. Cut the end of the pipette such that the distance from the beginning of the taper to the end of the pipette is 1.04cm to 1.05cm. The length of the pipette is inversely proportional to the caliber of the pipette orifice. Be aware that making a longer pipette a…

Discussion

Over 50 years ago, Billingham, Brent, and Medawar used in utero transplantation in mice to induce immune tolerance to foreign proteins.16 Since that time, several variations of this technique have been used to address questions in immunology and stem cell biology.

The protocol detailed here is one of the most accessible methods for IUT. The fetal liver offers an easily visualized target and provides access to the systemic circulation via the portal and hepatic veins. Ho…

Disclosures

The authors have nothing to disclose.

Acknowledgements

We would like to acknowledge our funding sources: The California Institute for Regenerative Medicine Clinical Fellow Training Grant (AN), National Science Foundation (MW), Irene Perstein Award (TCM), American College of Surgeons (TCM), American Pediatric Surgical Association (TCM), and the March of Dimes (TCM).

Materials

Material Name Type Company Catalogue Number Comment
Pipettes   Kimble 71900-100  
Pipette puller   Sutter Instruments Company Model P-30  
Microinjector   Narishige IM-300  
Pipette sharpener   Sutter Instruments Company Model BV-10  

References

  1. Flake, A. W. Treatment of X-linked severe combined immunodeficiency by in utero transplantation of paternal bone marrow. N Engl J Med. 335, 1806-1810 (1996).
  2. Wengler, G. S. In-utero transplantation of parental CD34 haematopoietic progenitor cells in a patient with X-linked severe combined immunodeficiency (SCIDXI). Lancet. 348, 1484-1487 (1996).
  3. Flake, A. W., Zanjani, E. D. in utero hematopoietic stem cell transplantation: ontogenic opportunities and biologic barriers. Blood. 94, 2179-2191 (1999).
  4. Merianos, D. J. Maternal alloantibodies induce a postnatal immune response that limits engraftment following in utero hematopoietic cell transplantation in mice. J Clin Invest. 119, 2590-2600 (2009).
  5. Peranteau, W. H., Endo, M., Adibe, O. O., Flake, A. W. Evidence for an immune barrier after in utero hematopoietic-cell transplantation. Blood. 109, 1331-1333 (2007).
  6. Kim, H. B., Shaaban, A. F., Yang, E. Y., Liechty, K. W., Flake, A. W. Microchimerism and tolerance after in utero bone marrow transplantation in mice. J Surg Res. 77, 1-5 (1998).
  7. Durkin, E. T., Jones, K. A., Rajesh, D., Shaaban, A. F. Early chimerism threshold predicts sustained engraftment and NK-cell tolerance in prenatal allogeneic chimeras. Blood. 112, 5245-5253 (2008).
  8. Mackenzie, T. C., Shaaban, A. F., Radu, A., Flake, A. W. Engraftment of bone marrow and fetal liver cells after in utero transplantation in MDX mice. J Pediatr Surg. 37, 1058-1064 (2002).
  9. Hayashi, S. Mixed chimerism following in utero hematopoietic stem cell transplantation in murine models of hemoglobinopathy. Exp Hematol. 31, 176-184 (2003).
  10. Bouchard, S. Long-term transgene expression in cardiac and skeletal muscle following fetal administration of adenoviral or adeno-associated viral vectors in mice. J Gene Med. 5, 941-950 (2003).
  11. Meza, N. W. Rescue of pyruvate kinase deficiency in mice by gene therapy using the human isoenzyme. Mol Ther. 17, 2000-2009 (2009).
  12. MacKenzie, T. C. Efficient transduction of liver and muscle after in utero injection of lentiviral vectors with different pseudotypes. Mol Ther. 6, 349-358 (2002).
  13. MacKenzie, T. C. Transduction of satellite cells after prenatal intramuscular administration of lentiviral vectors. J Gene Med. 7, 50-58 (2005).
  14. Sabatino, D. E. Persistent expression of hF.IX After tolerance induction by in utero or neonatal administration of AAV-1-F.IX in hemophilia B mice. Mol Ther. 15, 1677-1685 (2007).
  15. Mellor, A. L., Munn, D. H. Immunology at the maternal-fetal interface: lessons for T cell tolerance and suppression. Annu Rev Immunol. 18, 367-391 (2000).
  16. Billingham, R. E., Brent, L., Medawar, P. B. Actively acquired tolerance of foreign cells. Nature. 172, 603-606 (1953).
  17. Endo, M. Gene transfer to ocular stem cells by early gestational intraamniotic injection of lentiviral vector. Mol Ther. 15, 579-587 (2007).
  18. Waddington, S. N. Long-term transgene expression by administration of a lentivirus-based vector to the fetal circulation of immuno-competent mice. Gene Ther. 10, 1234-1240 (2003).
  19. Schachtner, S., Buck, C., Bergelson, J., Baldwin, H. Temporally regulated expression patterns following in utero adenovirus-mediated gene transfer. Gene Ther. 6, 1249-1257 (1999).
check_url/2303?article_type=t

Play Video

Cite This Article
Nijagal, A., Le, T., Wegorzewska, M., MacKenzie, T. C. A Mouse Model of in Utero Transplantation. J. Vis. Exp. (47), e2303, doi:10.3791/2303 (2011).

View Video